5,031
Views
81
CrossRef citations to date
0
Altmetric
Research Articles

Fever, hyperthermia and the heat shock response

&
Pages 423-435 | Received 01 Apr 2013, Accepted 21 May 2013, Published online: 17 Jul 2013

Abstract

The heat shock response is a highly conserved primitive response that is essential for survival against a wide range of stresses, including extremes of temperature. Fever is a more recently evolved response, during which organisms raise their core body temperature and temporarily subject themselves to thermal stress in the face of infections. The present review documents studies showing the potential overlap between the febrile response and the heat shock response and how both activate the same common transcriptional programme (although with different magnitudes) including the stress-activated transcription factor, heat shock factor-1, to modify host defences in the context of infection, inflammation and injury. The review focuses primarily on how hyperthermia within the febrile range that often accompanies infections and inflammation acts as a biological response modifier and modifies innate immune responses. The characteristic 2–3 °C increase in core body temperature during fever activates and utilises elements of the heat shock response pathway to modify cytokine and chemokine gene expression, cellular signalling and immune cell mobilisation to sites of inflammation, infection and injury. Interestingly, typical proinflammatory agonists such as Toll-like receptor agonists modify the heat shock-induced transcriptional programme and expression of HSP genes following co-exposure to febrile range hyperthermia or heat shock, suggesting a complex reciprocal regulation between the inflammatory pathway and the heat shock response pathway.

Introduction

Fever is a complex physiological response to infection and injury, the key feature of which is a temporary resetting of the body's thermostatic set point resulting in an increase in core temperature. Although fever is recognised as a component of the acute-phase response to infection and perceived to be a response limited to mammals and birds, many poikilothermic animals, including lower vertebrates, arthropods, and annelids, also increase their core temperature in response to infection or injury [Citation1]. The prevalence of fever in such diverse modern animals suggests that it first appeared over 600 million years ago. This evolutionary persistence of fever is even more remarkable when one considers its substantial metabolic cost. In humans, generating fever through thermogenic shivering requires up to a 6-fold increase in metabolic rate [Citation2], and maintaining a physiological core temperature at febrile levels requires an approximately 12% increase in metabolic rate per 1 °C increase in core temperature [Citation3,Citation4]. In poikilothermic animals with infections, moving to warmer environs not only requires increased energy expenditure, but may also expose vulnerable individuals to attack by predators. Therefore, fever must confer benefit that generally outweighs these costs in the infected or injured host. Furthermore, given the phylogenetic age of fever, the immunological processes that are active during febrile illnesses have had ample opportunity to evolve for optimal function at febrile temperatures.

This review will focus on how the characteristic 2–3 °C increase in body temperature that often accompanies infections and inflammation acts as a biological response modifier by regulating signalling pathways and gene expression involved in immune defence, inflammation, and cell death and survival. We discuss how elements of the heat shock (HS) response pathway have been co-opted as immune response modifiers and how the knowledge of how the temperature responsiveness of elements of the immune response can be translated to the care of the acutely ill patient.

Heat shock response

While fever is a systemic response to infection and injury, the HS response acts as a defence mechanism against cellular stress. The HS response, a highly conserved ancient biological process, is essential for survival against a myriad of environmental stresses, including extremes of temperature, chemicals and radiations, each of which can cause denaturation of essential cellular proteins. Also referred to as the ‘cellular stress response’ the HS response is accompanied with reprogramming of the cellular transcriptional and translational machinery to preferentially express a set of stress-inducible proteins namely the heat shock proteins (HSPs). During stress these HSPs act as chaperones and bind to denatured proteins to either preserve them until the stress has abated or to target the denatured proteins for degradation [Citation5–7]. Genes encoding the five families of HSPs are highly conserved. Their presence in all species studied to date including archaebacteria, eubacteria, and eukaryotes, suggests that they first arose at least 2.5 billion years ago. While prokaryotic and eukaryotic HSP genes exhibit striking cross-domain homology, they use different mechanisms of transcriptional regulation. In eukaryotes, HSP expression is regulated at the transcriptional level by the stress-activated transcription factor heat shock factor (HSF). Mammals, including humans have three HSF orthologues of which HSF1 is the heat inducible orthologue [Citation8–10]. Human HSF1 is a complex protein with an N-terminal DNA binding domain, three hydrophobic regions that regulate trimerisation, a serine-rich regulatory domain that regulates trasnscriptional activation, and two independent C-terminal transactivation domains [Citation11]. HSF1 is retained as inactive monomers by intramolecular interactions between the first two hydrophobic regions and the third hydrophobic region [Citation12,Citation13]. During HSF1 trimerisation, the intramolecular interactions are replaced by intermolecular interactions between the first two hydrophobic repeats of each of the three trimerising HSF1 molecules. Heat-inducible HSFs, including mammalian HSF1 exists in dynamic equilibrium between a transcriptionally inactive, cytosolic, hetero-oligomeric pool and a transcriptionally active intranuclear homotrimeric pool. Spontaneous trimerisation of HSF has been reported to be concentration-dependent, spontaneously forming DNA-binding trimers when present at sufficiently high concentrations in cell-free reactions [Citation12] and within intact cells [Citation13]. Zhong et al. [Citation14] demonstrated that dilution of trimerised Drosophila HSF in crude cell lysates from Schneider line-2 (SL-2) cells caused reversible dissociation of HSF trimers to monomers. They used this model system to calculate the equilibrium constant, Kd, for the HSF trimer dissociation reaction and showed that the Kd decreased, thereby favouring trimerisation, as the reaction temperature increased or upon exposure to oxidant stress. This analysis provides a useful conceptual model to interpret studies in which the expression levels of HSF may vary widely by describing the mathematical relationship among HSF concentration, temperature, and the extent of HSF trimerisation. Importantly, this study also demonstrates that HSF trimerisation may occur as a continuous temperature-dependent process that is activated over a temperature range rather than as a binary process activated when a distinct thermal threshold is exceeded.

Induction of HSF1 trimerisation is a hierarchical process. HSF is directly activated by heat in cell-free reactions, but the temperature range at which the reaction occurs is a species-specific intrinsic property of HSF, and related to the normal temperature range of the organism. For example, cell-free Drosophila HSF undergoes trimerisation between 28° and 38 °C while mouse HSF1 trimerises between 37° and 39 °C [Citation14,Citation15]. While cell-free HSF can be directly activated by heat, Zhong et al. showed that trimerisation of intracellular HSF is heat-activated at lower temperatures than cell-free HSF or by chemical stresses that have no effect on cell-free HSF, such as salicylate, dinitrophenol, arsenite, and ethanol [Citation14]. When human HSF1 is expressed in Drosophila SL-2 cells, it trimerises at 32–37 °C, the Drosophila HS range, rather than the usual human temperature threshold [Citation16]. In addition, the temperature threshold for mammalian HSF1 can differ between different tissues in the same organism [Citation17] and change over time such as after prolonged exposure to hyperthermia [Citation18] or in response to soluble mediators like arachidonic acid or type I interferon [Citation19,Citation20].

Activation of HSF1 and its transcriptional activity is greatly dependent upon its post-transcriptional modifications. HSF1 has 60 serines and threonines, at least 12 of which have been shown to be phosphorylated [Citation21] by various kinases including members of the MAP kinase pathway. Most of the phosphorylation events modify trans-activation domain function but phosphorylation of threonine 142 increases [Citation22] and phosphorylation of serine-121 reduces [Citation23] activation of human HSF1 to its DNA binding trimeric form and phosphorylation of serine-419 is required for its heat-induced nuclear translocation independent of trimerisation [Citation24]. HSF1 undergoes additional covalent modifications including sumoylation that are critical for its transcriptional activation of HSP genes [Citation25,Citation26]. Collectively, these studies demonstrate that HSF1 trimerisation and its transcriptional competency is regulated through multiple steps, each affected by temperatures, soluble mediators, and protein modifying signalling events that are encountered during febrile illnesses.

Fever and the HS response

Over twelve years ago we proposed a partial overlap between fever and the HS response [Citation27] based on data showing partial activation of HS signalling pathways at febrile temperatures, the participation of HSF1 in the regulation of several inflammatory mediator genes, and the cytoprotective effects of intracellular HSPs generated at febrile temperatures. In the subsequent decade additional experimental evidence has been generated that supports a functional overlap between fever and the HS response and identifies HSF1 as central to the relationship between these two distinct, evolutionarily conserved host defence mechanisms.

HSF1 activation has not only been shown to occur at temperatures in the febrile range but the temperature required for HSF1 activation and HSP gene expression has been shown to differ across species [Citation28] and across different cell types and tissues in the same organism [Citation17,Citation29], and to be lowered by exposure to certain inflammatory mediators [Citation19,Citation20]. For example, mouse lymphoid tissues, including spleen, exhibit a low thermal threshold for induction of HSP expression, which appears to derive from T lymphocyte rather than B lymphocyte behaviour [Citation29,Citation30], suggesting variable cell- and tissue-specific activation of HS response at febrile temperatures ().

Table I. HSF1 activation and HSP induction in various tissue and tissue culture cells at febrile range temperatures.

As discussed in the previous section, HSF1 trimerisation and nuclear translocation is required but not sufficient for gene transcription [Citation12,Citation13] and is dissociable from HSP gene transcription [Citation31,Citation32]. We found that exposing the RAW 264.7 mouse macrophage cell line to hyperthermia in the febrile range (39.5 °C) for a brief period activates HSF1 trimerisation and DNA binding activity, but is insufficient to induce expression of HSP genes, while exposing the cells to classic HS temperatures (≥42 °C) induces high levels of Hsp70 [Citation33]. Similarly, Laszlo et al. [Citation34] showed that 15 min exposure to 38 °C was sufficient to activate HSF1 to a DNA binding form in HA-1 hamster fibroblasts and C3H10T1/2 mouse fibroblast-like cells. We recently confirmed that HSF1 activation to its DNA-binding trimeric state is dissociable from Hsp70 expression and showed that the thermal threshold for Hsp70 expression is both temperature- and time-dependent in the A549 human pulmonary epithelial-like adenocarcinoma cell line [Citation35]. Similar to the results in the Laszlo study, exposing A549 cells to 38.5 °C, 39.5 °C, and 41 °C for 1 h each caused similar nuclear translocation and DNA binding activity of HSF1. However, detectable Hsp70 protein expression required 24 h exposure at 38.5 °C, 6 h exposure at 39.5 °C, and only 1 h exposure at 41 °C (). The relationship between the exposure temperature and maximal Hsp70 protein levels was linear between 37 °C and 41 °C, increasing approximately 50%/ °C. However, a further 1 °C increase in temperature to 42 °C stimulated an additional 2.6-fold increase in Hsp70 expression with little additional activation of HSF1 binding activity () [Citation35]. These results suggest that 41–42 °C may represent a key temperature threshold in human cells above which the relationship between Hsp70 gene activation and temperature shifts. That 41 °C is the upper limit of the normal human febrile range underscores the biological significance of this relationship [Citation36,Citation37].

Figure 1. Hsp72 protein expression is temperature and time dependent. Subconfluent A549 monolayers were exposed to the indicated temperature for the indicated time and then were switched to 37 °C for the remainder of a 24-h incubation. Cells were lysed and analysed for Hsp72 levels by immunoblotting. (A) Band intensities were analysed by direct imaging of the chemiluminescent signal, corrected for loading by normalising to β-tubulin levels, and standardised to 37 °C baseline levels (0). (B) Hsp72 protein levels after 6 h exposure to the indicated temperature between 38.5 °C and 41 °C or to 42 °C for 2 h followed by 4 h recovery at 37 °C were compared. Data are mean ± SE of six experiments. *p < 0.05 versus time 0. †p < 0.05 and ¶p < 0.05 versus 38.5 °C and 39.5 °C, respectively, values at the same exposure time. This research was originally published in Cell Stress and Chaperones [Citation35]. Reprinted with kind permission from Springer Science and Business Media.

Figure 1. Hsp72 protein expression is temperature and time dependent. Subconfluent A549 monolayers were exposed to the indicated temperature for the indicated time and then were switched to 37 °C for the remainder of a 24-h incubation. Cells were lysed and analysed for Hsp72 levels by immunoblotting. (A) Band intensities were analysed by direct imaging of the chemiluminescent signal, corrected for loading by normalising to β-tubulin levels, and standardised to 37 °C baseline levels (0). (B) Hsp72 protein levels after 6 h exposure to the indicated temperature between 38.5 °C and 41 °C or to 42 °C for 2 h followed by 4 h recovery at 37 °C were compared. Data are mean ± SE of six experiments. *p < 0.05 versus time 0. †p < 0.05 and ¶p < 0.05 versus 38.5 °C and 39.5 °C, respectively, values at the same exposure time. This research was originally published in Cell Stress and Chaperones [Citation35]. Reprinted with kind permission from Springer Science and Business Media.

In anaesthetised mice, raising core temperature to febrile-range levels (rectal temperature 39.5 °C) by partial immersion in a heated water bath for 3 h was sufficient to activate Hsp72 expression in liver and kidney, albeit at much lower levels than mice exposed to HS temperature (rectal temperature 42 °C) for only 20 min followed by 160 min normothermic recovery [Citation38]. More recently, we showed that maintaining core temperature at 39.5 °C for 24 h in conscious mice activates expression of Hsp70 in lung parenchyma [Citation39,Citation40]. The relatively low temperature-dependent expression of Hsp70 at temperatures within the normal febrile temperature range and the profound increase in Hsp70 expression at temperatures ≥42 °C supports our proposition that fever and HS responses are distinct but partially overlapping processes [Citation27].

Fever, inflammation and immune responses

As expected based on the evolutionary conservation of the febrile response [Citation1,Citation27,Citation41], fever and hyperthermia in the febrile-range (febrile range hyperthermia (FRH), core temperature ∼39.5 °C) confers protection in infection by improving pathogen clearance in vivo [Citation39,Citation42–44] although the change in temperature have little effect on the growth rate of the pathogens [Citation39,Citation44]. Increasing body temperature of fish and lizards by ∼4 °C greatly increased clearance of the same Gram-negative pathogen, Aeromonas hydrophila, despite a 10 °C lower temperature range in the fish [Citation42,Citation43]. These data demonstrate that FRH enhances pathogen clearance in vivo through effects on host defence rather than on the pathogen. This effect may also explain the association of fever with improved survival in retrospective clinical studies of bacterial infections [Citation45–47].

We developed a mouse model of FRH in which mice exposed to an ambient temperature of 36–37 °C increase their core temperature by 2–3 °C but maintain normal circadian patterns and appear otherwise healthy and active [Citation48,Citation49]. Using this model we showed that FRH accelerated pathogen clearance in experimental Klebsiella pneumoniae peritonitis [Citation44] and pneumonia [Citation39]. Focusing on the lung, we found that FRH, despite reducing pathogen load, tended to reduce survival in the K. pneumoniae model while it greatly improved survival in the peritonitis model [Citation39,Citation44] and severe lung injury was found in mice co-exposed to FRH in the pneumonia model [Citation39,Citation44]. Lipke and Martin et al. confirmed these results in the intratracheal bacterial lipopolysaccharide (LPS)-challenged mouse model [Citation50,Citation51]. Co-exposure to FRH and LPS exerted similar effects in a model of lethal pulmonary oxygen toxicity [Citation52]. Considering these results it appears that FRH augmented innate immune processes, which accelerated pathogen clearance but also enhanced collateral tissue injury, and the net effect on survival depended on the balance between the two effects. Although the above studies focused primarily on the host’s inflammatory responses and neutrophil-mediated vascular injury, the contribution of other factors cannot be ignored. For example, using a similar mouse model of lung injury, D’Alessio et al. [Citation53] identified a critical role for regulatory T cells (Tregs) in resolution of lung injury and showed that depletion/absence of Tregs prolonged LPS-induced proinflammatory responses, reduced neutrophil apoptosis and severely delayed recovery. Considering that hyperthermia and HSPs both greatly modify T cell behaviour [Citation54–57], the contribution of dysregulated Treg mechanisms in lung injury at FRH could not be negated. However, few studies have focused on this aspect of hyperthermia and inflammation.

FRH exposure greatly increased neutrophil infiltration in both the pulmonary oxygen toxicity and intratracheal LPS instillation models [Citation39,Citation52]. In fact, the studies showed that exposure to FRH augmented multiple steps required for neutrophil delivery to sites of infection and injury, including induction of G-CSF expression and expansion of the circulating neutrophil pool [Citation58], increased generation of the CXC chemokine family of endogenous chemotaxins [Citation39,Citation49], and increased capacity for chemokine-directed transendothelial migration (TEM) of neutrophils [Citation59,Citation60]. Furthermore, adoptive transfer of fluorescently labelled neutrophils between normothermic and hyperthermic neutrophil donors and recipients demonstrated that enhanced neutrophil migration capacity required FRH exposure of both the donors and recipients indicating that FRH augments neutrophil transmigration capacity through interdependent effects on both the neutrophils and the vascular endothelia [Citation59].

In addition to augmented neutrophil accumulation in lung, FRH co-exposure had two additional effects in the LPS-challenged mouse lung that are also characteristic of human acute respiratory distress syndrome (ARDS), endothelial hyperpermeability and epithelial injury. In the mouse intratracheal LPS instillation model, exposure to FRH caused extensive epithelial injury [Citation39], and Lipke et al. [Citation50,Citation51] showed it to be caused by augmented TNFα- and fas-dependent apoptosis. Using human neutrophils and the mouse MLE15 lung epithelial cell line, we found that exposure to 39.5 °C greatly accelerates activation of all three initiator caspases, caspase-2, 8, and 10, with evidence of activation as early as 60 min after stimulation with TNFα or agonistic anti-fas antibody (and within 30 min if treated at 42 °C) [Citation61,Citation62]. Accelerated and augmented apoptosis in the FRH-exposed and TNFα- or anti-fas-treated cells was partially blocked by inhibition of all three initiator caspases, did not require HSF1, and still occurred even when NFκB activation was independently blocked by expression of the IκBα super-repressor [Citation62].

The Evans and Repasky laboratories have shown many of the same effects of FRH on cytokine gene expression [Citation63,Citation64], but have extended their studies of FRH effects to lymphocyte trafficking [Citation65–67]. Utilising intravital microscopy to analyse lymphocyte trafficking in high endothelial venules in mice, the Evans laboratory has shown that exposing lymphocytes to FRH enhances their L-selectin- and alpha4beta7 integrin-dependent binding to high endothelial venules that increased their trafficking to secondary lymphoid tissue [Citation65,Citation66] and the enhanced migration is mediated through the IL-6-dependent endothelial expression of ICAM-1 [Citation57]. The consequences for the FRH-enhanced lymphocyte recruitment in infections and inflammatory disease have not yet been demonstrated experimentally. Recently, Lee et al. [Citation63] showed a unique effect of FRH on activation and reprogramming of macrophages whereby FRH exposure causes a transitory reduction in endotoxin tolerance behaviour in vivo, increases inflammatory macrophage recruitment and maintains a sustained responsiveness to LPS.

Collectively, these studies demonstrate that exposure to hyperthermia achieved during febrile illness has many effects on gene expression, cell signalling, and cell behaviour that includes leucocyte and macrophage recruitment, opening of endothelial paracellular pathways to macromolecules and enhance extrinsic apoptosis in epithelium. These effects can be both beneficial and harmful, and the consequence for host survival and recovery depend on the nature of the pathological process.

HSF1: the central mediator

Heat-inducible HSF, including mammalian HSF1, was originally identified as a stress-activated transcriptional activator of HSP genes. However, evidence for the participation of HSF in more diverse processes such as innate immunity in Caenorhabditis elegans [Citation68] and extra-embryonic development [Citation69] suggest a much broader range of biological functions than previously thought.

The concept that HSF1 might have additional functions was initially suggested by Westwood et al. [Citation70] who used in situ hybridisation analysis to show that HS stimulated the recruitment of HSF to 150 distinct chromosomal loci in Drosophila salivary gland polytene chromosomes, far more than could be accounted for by the known HSP genes. These observations were subsequently complemented by Trinklein et al. [Citation71] who used a combination of chromatin immunoprecipitation and human promoter microarray analyses to show recruitment of HSF1 to multiple non-HSP genes in human K562 cells. Our own in silico analysis of CXC chemokine genes showed that the promoter regions of almost all mouse and human CXC chemokine genes contained multiple potential HSE consensus sequences [Citation72]. We subsequently showed that some of the putative HSEs recruited HSF1 in vivo and that some of these functioned as a transcriptional activator, some as a repressor and some were functionally silent [Citation73,Citation74]. Additional studies, using cDNA microarrays to analyse the gene expression pattern activated by HS confirmed that exposure to HS also modifies expression of several non-hsp genes, including those involved in regulation of transcription, growth, DNA repair, apoptosis, signalling, and cytoskeletal function [Citation75–77]. More recently, Mendillo et al. [Citation78] showed that HSF1 was activated under basal conditions in cancers with high tumorigenic and metastatic potential but not in other cancers. Using high throughput ChIP-sequencing, they showed that HSF1 was recruited to about 500 genes many of which are distinct from those induced by HS and some of which are down-regulated by HSF1.

Studies with the HSF1 knock-out mouse also confirmed HSF1 as the major regulator of the heat/stress response [Citation9], but also demonstrated its participation in the regulation of extra-embryonic development, growth, and endotoxaemia-induced systemic inflammation [Citation69], female [Citation69,Citation79] and male [Citation80] reproductive potential, the ubiquitin proteolytic pathway [Citation81], post-natal brain development [Citation82], in the maintenance of olfactory epithelium and in ciliary beating in the respiratory epithelium, ependymal cells, oviduct, and the trachea [Citation83,Citation84], and a potent promoter of tumorigenesis [Citation85].

Gene-specific studies by our laboratory and by Stuart Calderwood’s laboratory have shown that HSF1 can modify the expression of various cytokines, chemokines and acute response genes. The Calderwood group showed that following HS, HSF1 mediates transcriptional repression of human pro-interleukin-1β, c-fms, and c-fos genes [Citation86–89] through quenching of participating trans-activating factors, most notably NF-IL6/c/EBPβ. In our studies we found that HSF1 was activated at febrile-range temperatures (39.5 °C) and mediated the repression of TNFα gene expression by interacting with a putative HSE sequence present in the mouse TNFα 5'-untranslated region [Citation33,Citation90,Citation91]. Interestingly, we found that exposure to febrile-range temperatures also represses TNFα gene expression by selectively blocking recruitment of NFκB and Sp1 to the TNFα proximal promoter sequence [Citation92,Citation93]. In further support of HSF1 as a negative regulator of TNFα expression, HSF1-null mice exhibit higher circulating levels of TNFα expression after intraperitoneal challenge with LPS [Citation69] and higher levels of TNFα in lung lavage after intratracheal LPS challenge [Citation93]. In addition, activated HSF1 has also been found to repress human CXCL5 [Citation73] and the pro-apoptotic factor, xIAP-associated factor 1 [Citation94].

Regarding HSF1-mediated induction of non-HSP genes, we analysed the effect of HS on the expression of interleukin IL-8 [Citation74]. HS enhanced TNFα-induced IL-8 secretion in human A549 epithelial cells but unlike classic HSPs, HS alone was not sufficient to activate IL-8 expression. Using EMSA and ChIP, we identified two IL-8 promoter regions, 800 and 1200 nt upstream of the transcription start site, that bound active HSF1 and, using a 5’-deletion mapping strategy and siRNA knockdown of HSF1, we showed that the interaction of HSF1 with both promoter regions contributed to the increased IL-8 expression in cells co-treated with TNFα and HS. Goldring et al. [Citation95] found that activated HSF1 has a similar co-activator function for the murine iNOS gene. Inouye et al. [Citation96] found that HSF1 constitutively bound to IL-6-associated chromatin in unstressed peritoneal macrophages and fibroblasts and enhanced LPS-induced IL-6 expression by modifying the chromatin accessibility of other transcription factors. The Santoro lab reported two new non-classical HS genes, cyclooxygenase-2 and the zinc finger AN1-type domain-2a gene (AIRAP) that exhibit HS-inducible transcription similar to those of canonical HSPs [Citation97,Citation98].

HSF1 may also exert additional biological effects by binding to and modifying function of proteins involved in diverse cellular processes, including HSPs [Citation11], the nuclear pore-forming TPR protein through which Hsp72 is secreted [Citation99], the catalytic subunit of the DNA-dependent protein kinase [Citation100], other transcription factors [Citation86,Citation101,Citation102], components of the TFIIB transcription complex [Citation103], the cell division cycle protein, Cdc20 [Citation104], the apoptosis modulator DAXX [Citation105], and the multidrug exporter, RalBP [Citation106]. Collectively, these studies illustrate the broad range of important biological functions of HSF1 and underscore the potential importance and impact of altered HSF1 expression levels and genetic variations in the host cell.

HSF1 genetic variations and potential consequences

As discussed in the previous sections, HSF1 regulates expression of a broad range of genes, including those involved in host defence, inflammation and tumorigenesis as well as exerting additional effects by directly binding to proteins critical for cell proliferation, survival, and death. Studies from heterozygous mice suggest that the level of HSF1 expression may affect capacity for expression of some chemokines [Citation74] and modify risk of tumour progression [Citation85]. Considering the central participation of HSF1 in so many important biological functions, it is surprising that so little is known about genetic variations in elements of the human HS response, especially HSF1, and the potential impact on human health and disease.

Although single-nucleotide polymorphisms (SNPs) have been identified in various HSP genes, few studies have focused on the HSF1 molecule. Recently, Li et al. [Citation107] reported two novel SNPs in the HSF1 gene that are disproportionately associated with thermal tolerance in Chinese Holstein cattle, including one 3’UTR SNP that disrupts a potential microRNA binding sequence in HSF1. However, the occurrence of SNPs in the human HSF1 gene has not yet been systematically analysed. To begin to understand the potential biological and clinical importance of SNPs in the HSF1 gene we analysed the human HSF1 gene for SNPs by mining the NCBI dbSNP database and performing exonic sequencing from anonymous genomic DNA samples. DNA was isolated from 30 healthy Caucasians and 30 healthy African American volunteers, exons amplified by PCR, and bidirectional sequencing performed and each sequence was compared with a reference human HSF1 sequence (NT_037704). Mining the dbSNP database revealed six SNPs (three in the 3′UTR and three in the coding sequence). One of the coding SNPs caused a proline-to-threonine missense at amino acid 365 adjacent to LZ3 and one caused a frame-shift replacement of the 26-amino acid C-terminal transactivation domain. Direct sequencing confirmed the P365T SNP and identified two novel 5’UTR and two novel 3′UTR SNPs [Citation108]. Four of the five 3′UTR SNPs alter predicted miRNA target sequences as identified using the MicroSNiPer online program [Citation109] and both of the 5′UTR SNPs alter the 5’UTR secondary structure predicted using the RNAFold online program [Citation110]. The frequency of these and potentially other HSF1 SNPs and their participation in disease pathophysiology are not yet known.

HS response during infection and sepsis

Modifications in HSF1 activation and HSP expression have been demonstrated in clinical studies of human infections and in experimental infections in animals (). The clinical studies of HS response in human sepsis are small and utilise different methods but generally show that HSP expression is higher in patients with sepsis. Hashiguchi et al. [Citation111] analysed Hsp27, 60, 72 and 90 expression levels in blood neutrophils using mean fluorescence intensity from flow cytometry in 21 patients with early sepsis and 14 healthy controls. They found that neutrophils from the patients with sepsis had higher levels of all four HSPs compared with the control subjects. Similarly, Delogu et al. [Citation112] compared the proportion of peripheral blood mononuclear cells expressing Hsp72 by flow cytometry to be almost 4-fold higher in patients with sepsis than in healthy controls. Other studies showed that levels of cell-free Hsp72 in serum were also elevated in patients with sepsis, including children with septic shock [Citation113–115].

Table II. HSF1 activation and HSP expression in infections and injury.

Most studies of animal models of infections also demonstrate increased expression of host HSPs associated with the infection. In mice infected with Francisella tularensis, the pathogen causing tularaemia, peritoneal macrophages exhibited increased Hsp72 levels but not until day three of the infection [Citation116]. Trichinella infection in rats is associated with increased levels of Hsp25, 60, and 72 protein in spleen and brain, increased Hsp25 protein levels in liver, and increased Hsp25 and 60 levels in muscle [Citation117]. On the other hand, Singleton et al. [Citation118] reported reduced Hsp25 and 72 expression in lung 24 h after sepsis induced by cecal ligation and puncture. Weiss et al. [Citation119] reported that Hsp72 mRNA and protein levels in lung did not increase up to 48 h after cecal ligation and puncture in mice. In the colonic epithelium of mice, steady-state levels of Hsp72 and 25 are constitutively maintained by commensal bacteria through MyD88-mediated TLR signalling and these HSPs play a crucial role in the maintenance of intestinal epithelial homeostasis [Citation120]. Collectively, these data suggest the observed effect of infections on HSP expression may depend on timing relative to infection, the type of infection, and the tissue studied.

HSP induction is not limited to bacterial infections as several viruses also induce HSP expression in target cells, including Epstein-Barr virus in human B lymphocytes [Citation121], respiratory syncitial virus in A549 cells [Citation122], and adenovirus in B16 melanoma cells [Citation123]. HS response activation has also been found to occur in non-infectious inflammatory disorders, including human pancreatitis in which HSF1 was found to be activated [Citation124] and a mouse model of cerulean-induced pancreatitis in which elevated pancreatic levels of Hsp72 and 25 protein and activated HSF1 were detected [Citation125]. In their study of HSP expression in the mouse cecal ligation and puncture, Weiss et al. [Citation119] found elevated Hsp72 mRNA and protein levels in lungs of mice undergoing sham cecal ligation and puncture in which an abdominal incision was made. These results indicate that the stress of surgery might be sufficient to activate the HS response in mice as has been shown to occur in human cardiac surgery in which cardiac expression of Hsp72 was detected post-operatively [Citation126,Citation127].

Extracellular HSPs

Within the past decade HSPs have been shown to have additional cellular functions directly related to inflammation and the innate immune response. HSPs, particularly Hsp70, have been detected in the extracellular milieu and have been reported to be pro-inflammatory agonists for TLR2 and TLR4 [Citation128–130]. Although some earlier studies raised concerns that the TLR4 agonist activity of recombinant Hsp70 preparations was caused by LPS contamination [Citation131,Citation132], subsequent studies showing activity in recombinant Hsp70 generated in insect cells and non-recombinant Hsp70 as well as classic LPS controls support proinflammatory TLR4 agonist and macrophage activating activities of Hsp70 protein itself [Citation133–137]. In contrast to these reports, there is also strong evidence suggesting a potent anti-inflammatory role of HSPs that includes regulation of T cell responses, reducing stimulatory capacity of dendritic cells, and inducing development of immunosuppressive Treg cells [Citation138–141]. While the exact role of exogenous/extracellular HSPs is still debated, it is likely that HSPs can stimulate both innate and adaptive immune responses, at least in the context of infection and fever. This might help explain, at least in part, the molecular mechanisms by which fever and hyperthermia modify host responses in the face of severe infection and how dysregulated responses could lead to severe sepsis and multi-organ dysfunction.

Not only the functional role of extracellular HSPs but also its release mechanism is poorly understood. HSPs lack a classical consensus signal required for secretion and HSP secretion is not blocked by typical inhibitors of the endoplasmic reticulum–Golgi pathway, such as brefeldin A [Citation142]. In the initial reports by Hightower and Guidon [Citation143] and later by Hunter-Lavin et al. [Citation142], both groups showed that Hsp70 release occurred from healthy uninjured cells independent of cell death. Basu et al. [Citation144] showed that bioactive Hsp70 was also released from necrotic cells but not from cells undergoing apoptosis, and Mambula et al. [Citation145] showed that Hsp70 was released from prostate cancer cells via both necrosis and active secretion. Collectively, these studies suggest that Hsp70 is released actively by a non-classical secretory pathway and passively as a result of cellular necrosis but not apoptosis. To account for its active release in the absence of a leader sequence, several mechanisms have been proposed, including release by secretory-like granules [Citation146], via membrane export vesicles [Citation135] and via a lysosome–endosome pathway, where Hsp70 translocates into lysosomes via an ATP binding cassette (ABC) transport-like system and is then exported from the cell via the endocytic process [Citation147,Citation148].

HS response and TLR agonists

While HSPs can activate TLR signalling, recent in vitro and in vivo studies suggest that TLR agonists, particularly LPS, can activate expression of HSPs in mammalian cells. Edelman et al. [Citation149] reported that LPS activated expression of Hsp60 and 70 in isolated rat lung pericytes at 37 °C, but the increase was modest, only 20 to 40%, and required 18-h incubation with LPS. Similarly, Hirsh and coworkers [Citation150] showed that in vitro treatment of human neutrophils with LPS at 37 °C stimulated a rapid increase in the percentage of cells with detectable intracellular and surface expression of Hsp60 and 70 as detected by flow cytometry, but the increase in HSP protein expression level was not reported. Administration of LPS to rats in vivo caused up-regulation of Hsp72 in splenocytes [Citation151,Citation152] and increased levels of Hsp70 mRNA in lung and liver compared with rats previously subjected to an endotoxin tolerance protocol [Citation153].

The release of Hsp60 and 70 into the circulation during infection, inflammation, and trauma combined with its TLR agonist activity might suggest a positive feedback mechanism that could amplify inflammation. Recently, we found that co-exposure to TLR agonists synergises with exposure to febrile temperatures to greatly augment Hsp70 synthesis and secretion in the RAW 264.7 mouse macrophage cell line [Citation40]. The increase in HSP expression is mediated through a p38 MAP kinase-dependent signalling pathway leading to increased histone H3 phosphorylation and HSF1 recruitment to the Hsp70 chromatin. The mechanism by which co-exposure to TLR agonists and febrile temperatures increases Hsp70 secretion is not yet known, but occurred without evidence of cytotoxicity. Similar synergism between TLR agonists and febrile temperatures for Hsp70 expression and secretion were seen in IL-1β-stimulated human A549 cells in vitro and in an intratracheal LPS-challenge mouse model of acute lung injury in vivo () [Citation40]. In the latter model, the combination of FRH and intratracheal LPS stimulated an increase in Hsp70 protein levels in lung homogenates and in cell-free lung lavage fluid. Considering the pyrogenic action of TLR agonists, including Hsp70, we propose that the synergism between fever and TLR agonists for synthesis and release of Hsp70 promotes a vicious proinflammatory cycle that may contribute to the negative consequences of fever in high acuity disease ().

Figure 2. TLR agonists augment Hsp70 expression and release. A and B: RAW cells were incubated with 100 ng/mL LPS, 0.5 µg/mL Pam3CSK4 (Pam3C) or 12.5 µg/mL poly(IC) (pI:C) at 39.5 °C for 6 h (A), or were heat shocked at 42 °C for 2 h, recovered at 37 °C for 4 h (B), lysed, and immunoblotted for Hsp70 and β-tubulin. Lane 1 is the untreated 37 °C control. C and D: RAW cells were incubated with 0, 100, or 1000 ng/mL LPS at 37 or 39.5 °C for 6 h (C) or 24 h (D). Cell culture supernatants were collected and cleared by centrifugation, and Hsp70 was quantified by ELISA and presented as pg/mL. Data presented as the means ± SE (n = 4). * and † denote p < 0.05 versus similarly treated 37 °C cells and 39.5 °C cells with no LPS or hyperthermia-exposed cells, respectively. E and F: Mice implanted with intraperitoneal thermistors were housed at either 25 °C (normothermic, NT) or 36–37 °C (hyperthermic, HT) ambient temperature. For LPS exposure, mice were intratracheally instilled with LPS or sterile phosphate buffered saline (PBS) (control) and housed under normothermic or hyperthermic conditions for 24 h. The lungs were excised, and the homogenates were immunoblotted for Hsp70 and expressed as a ratio to β-actin (E), or lungs were lavaged and Hsp70 quantified by ELISA in the lavage fluid (F). Data are presented as means ± SE (n = 4). *, †, and § denote p < 0.05 versus PBS-treated NT controls, PBS-treated HT mice, and LPS-treated NT mice, respectively. This research was originally published in the Journal of Biological Chemistry [Citation40]. © The American Society for Biochemistry and Molecular Biology.

Figure 2. TLR agonists augment Hsp70 expression and release. A and B: RAW cells were incubated with 100 ng/mL LPS, 0.5 µg/mL Pam3CSK4 (Pam3C) or 12.5 µg/mL poly(IC) (pI:C) at 39.5 °C for 6 h (A), or were heat shocked at 42 °C for 2 h, recovered at 37 °C for 4 h (B), lysed, and immunoblotted for Hsp70 and β-tubulin. Lane 1 is the untreated 37 °C control. C and D: RAW cells were incubated with 0, 100, or 1000 ng/mL LPS at 37 or 39.5 °C for 6 h (C) or 24 h (D). Cell culture supernatants were collected and cleared by centrifugation, and Hsp70 was quantified by ELISA and presented as pg/mL. Data presented as the means ± SE (n = 4). * and † denote p < 0.05 versus similarly treated 37 °C cells and 39.5 °C cells with no LPS or hyperthermia-exposed cells, respectively. E and F: Mice implanted with intraperitoneal thermistors were housed at either 25 °C (normothermic, NT) or 36–37 °C (hyperthermic, HT) ambient temperature. For LPS exposure, mice were intratracheally instilled with LPS or sterile phosphate buffered saline (PBS) (control) and housed under normothermic or hyperthermic conditions for 24 h. The lungs were excised, and the homogenates were immunoblotted for Hsp70 and expressed as a ratio to β-actin (E), or lungs were lavaged and Hsp70 quantified by ELISA in the lavage fluid (F). Data are presented as means ± SE (n = 4). *, †, and § denote p < 0.05 versus PBS-treated NT controls, PBS-treated HT mice, and LPS-treated NT mice, respectively. This research was originally published in the Journal of Biological Chemistry [Citation40]. © The American Society for Biochemistry and Molecular Biology.

Figure 3. Model of how fever, inflammatory agonists, and Hsp70 interact to cause sepsis. Proposed model of sepsis in which LPS and fever initiate a positive feedback pathway through enhanced Hsp70 expression and release, and subsequent increased TLR activation, Hsp70 expression, and proinflammatory cytokine release. This research was originally published in the Journal of Biological Chemistry [Citation40], © the American Society for Biochemistry and Molecular Biology.

Figure 3. Model of how fever, inflammatory agonists, and Hsp70 interact to cause sepsis. Proposed model of sepsis in which LPS and fever initiate a positive feedback pathway through enhanced Hsp70 expression and release, and subsequent increased TLR activation, Hsp70 expression, and proinflammatory cytokine release. This research was originally published in the Journal of Biological Chemistry [Citation40], © the American Society for Biochemistry and Molecular Biology.

Conclusions

In the present review we have discussed how the host’s febrile response share components of the HS pathway to generate an optimal host defence during inflammation, infection and injury. It is evident that the three components of the HS response pathway, namely the stressor (temperature), the central activator (HSF1), and the final product, HSPs, have all evolved to perform additional functions beyond the typical cellular stress response. All three components have strong immunmodulatory effects that include mobilisation of immune cells, regulation of proinflammatory cytokine/chemokine gene expression and activation of both pro- and anti-inflammatory pathways. Interestingly, the regulation is mutual between HS response and the inflammatory pathways and reciprocated by proinflammatory agents as well, which either augment the temperature effect or directly activate the HSP gene transcription programme. It is apparent therefore, that a better understanding of the complex interaction between the HS response and the inflammatory pathway is critical not only for conditions where both pathways are activated such as infection, sepsis and multi-organ dysfunction but also for optimal exploitation of thermotolerance and therapeutic hyperthermia, where dysregulated inflammatory signalling could severely compromise the efficiency and final outcome of hyperthermia therapy.

Declaration of interest

This work was supported by US National Institutes of Health grants GM069431 (ISS) and GM066855, HL69057 and HL085256 (J.D.H.), and by Veterans Association Merit Review grants (J.D.H. and I.S.S.). The authors alone are responsible for the content and writing of the paper.

References

  • Kluger MJ, Kozak W, Conn CA, Leon LR, Soszynski D. The adaptive value of fever. Infect Dis Clin North Am 1996;10:1–20
  • Horvath SM, Spurr GB, Hutt BK, Hamilton LH. Metabolic cost of shivering. J Appl Physiol 1956;8:595–602
  • Schumacker PT, Rowland J, Saltz S, Nelson DP, Wood LD. Effects of hyperthermia and hypothermia on oxygen extraction by tissues during hypovolemia. J Appl Physiol 1987;63:1246–52
  • Manthous CA, Hall JB, Olson D, Singh M, Chatila W, Pohlman A, et al. Effect of cooling on oxygen consumption in febrile critically ill patients. Am J Respir Crit Care Med 1995;151:10–14
  • Feder ME, Hofmann GE. Heat-shock proteins, molecular chaperones, and the stress response: Evolutionary and ecological physiology. Annu Rev Physiol 1999;61:243–82
  • Lindquist S. The heat-shock response. Annu Rev Biochem 1986;55:1151–91
  • Morimoto RI, Kline MP, Bimston DN, Cotto JJ. The heat-shock response: Regulation and function of heat-shock proteins and molecular chaperones. Essays Biochem 1997;32:17–29
  • Akerfelt M, Vihervaara A, Laiho A, Conter A, Christians ES, Sistonen L, et al. Heat shock transcription factor 1 localizes to sex chromatin during meiotic repression. J Biol Chem 2010;285:34469–76
  • McMillan DR, Xiao X, Shao L, Graves K, Benjamin IJ. Targeted disruption of heat shock transcription factor 1 abolishes thermotolerance and protection against heat-inducible apoptosis. J Biol Chem 1998;273:7523–8
  • Pirkkala L, Nykanen P, Sistonen L. Roles of the heat shock transcription factors in regulation of the heat shock response and beyond. FASEB J 2001;15:1118–31
  • Singh IS, Shah NG, Almutairy EA, Hasday JD. Role of HSF1 in infectious disease. In: Pockley AG, Santoro MG, Calderwood SK, eds. Prokaryotic and Eukaryotic Heat Shock Proteins in Infectious Disease. Vol. 4. Dordrecht: Springer, 2009, p. 31
  • Farkas T, Kutskova YA, Zimarino V. Intramolecular repression of mouse heat shock factor 1. Mol Cell Biol 1998;18:906–18
  • Zuo J, Rungger D, Voellmy R. Multiple layers of regulation of human heat shock transcription factor 1. Mol Cell Biol 1995;15:4319–30
  • Zhong M, Orosz A, Wu C. Direct sensing of heat and oxidation by Drosophila heat shock transcription factor. Mol Cell 1998;2:101–8
  • Sarge KD, Zimarino V, Holm K, Wu C, Morimoto RI. Cloning and characterization of two mouse heat shock factors with distinct inducible and constitutive DNA-binding ability. Genes Dev 1991;5:1902–11
  • Clos J, Rabindran S, Wisniewski J, Wu C. Induction temperature of human heat shock factor is reprogrammed in a Drosophila cell environment. Nature 1993;364:252–5
  • Sarge KD. Regulation of HSF1 activation and HSP expression in mouse tissues under physiological stress conditions. Ann N Y Acad Sci 1998;851:112–16
  • Buckley BA, Owen ME, Hofmann GE. Adjusting the thermostat: The threshold induction temperature for the heat-shock response in intertidal mussels (genus Mytilus) changes as a function of thermal history. J Exp Biol 2001;204:3571–9
  • Jurivich DA, Sistonen L, Sarge KD, Morimoto RI. Arachidonate is a potent modulator of human heat shock gene transcription. Proc Natl Acad Sci USA 1994;91:2280–4
  • Morange M, Dubois MF, Bensaude O, Lebon P. Interferon pretreatment lowers the threshold for maximal heat-shock response in mouse cells. J Cell Physiol 1986;127:417–22
  • Guettouche T, Boellmann F, Lane WS, Voellmy R. Analysis of phosphorylation of human heat shock factor 1 in cells experiencing a stress. BMC Biochem 2005;6:4
  • Soncin F, Zhang X, Chu B, Wang X, Asea A, Ann Stevenson M, et al. Transcriptional activity and DNA binding of heat shock factor-1 involve phosphorylation on threonine 142 by CK2. Biochem Biophys Res Commun 2003;303:700–6
  • Wang X, Khaleque MA, Zhao MJ, Zhong R, Gaestel M, Calderwood SK. Phosphorylation of HSF1 by MAPK-activated protein kinase 2 on serine 121, inhibits transcriptional activity and promotes HSP90 binding. J Biol Chem 2006;281:782–91
  • Kim SA, Yoon JH, Lee SH, Ahn SG. Polo-like kinase 1 phosphorylates heat shock transcription factor 1 and mediates its nuclear translocation during heat stress. J Biol Chem 2005;280:12653–7
  • Hietakangas V, Anckar J, Blomster HA, Fujimoto M, Palvimo JJ, Nakai A, et al. PDSM, a motif for phosphorylation-dependent SUMO modification. Proc Natl Acad Sci USA 2006;103:45–50
  • Hong Y, Rogers R, Matunis MJ, Mayhew CN, Goodson ML, Park-Sarge OK, et al. Regulation of heat shock transcription factor 1 by stress-induced SUMO-1 modification. J Biol Chem 2001;276:40263–7
  • Hasday JD, Singh IS. Fever and the heat shock response: distinct, partially overlapping processes. Cell Stress Chaperones 2000;5:471–80
  • Tomanek L, Somero GN. Interspecific- and acclimation-induced variation in levels of heat-shock proteins 70 (hsp70) and 90 (hsp90) and heat-shock transcription factor-1 (HSF1) in congeneric marine snails (genus Tegula): Implications for regulation of hsp gene expression. J Exp Biol 2002;205:677–85
  • Gothard LQ, Ruffner ME, Woodward JG, Park-Sarge OK, Sarge KD. Lowered temperature set point for activation of the cellular stress response in T-lymphocytes. J Biol Chem 2003;278:9322–6
  • Ostberg JR, Kaplan KC, Repasky EA. Induction of stress proteins in a panel of mouse tissues by fever-range whole body hyperthermia. Int J Hyperthermia 2002;18:552–62
  • Cotto JJ, Kline M, Morimoto RI. Activation of heat shock factor 1 DNA binding precedes stress-induced serine phosphorylation. Evidence for a multistep pathway of regulation. J Biol Chem 1996;271:3355–8
  • Bharadwaj S, Hnatov A, Ali A, Ovsenek N. Induction of the DNA-binding and transcriptional activities of heat shock factor 1 is uncoupled in Xenopus oocytes. Biochim Biophys Acta 1998;1402:79–85
  • Singh IS, Viscardi RM, Kalvakolanu I, Calderwood S, Hasday JD. Inhibition of tumor necrosis factor-alpha transcription in macrophages exposed to febrile range temperature. A possible role for heat shock factor-1 as a negative transcriptional regulator. J Biol Chem 2000;275:9841–8
  • Laszlo A, Moros EG, Davidson T, Bradbury M, Straube W, Roti Roti J. The heat-shock factor is not activated in mammalian cells exposed to cellular phone frequency microwaves. Radiat Res 2005;164:163–72
  • Tulapurkar ME, Asiegbu BE, Singh IS, Hasday JD. Hyperthermia in the febrile range induces HSP72 expression proportional to exposure temperature but not to HSF-1 DNA-binding activity in human lung epithelial A549 cells. Cell Stress Chaperones 2009;14:499–508
  • Roth J, Rummel C, Barth SW, Gerstberger R, Hubschle T. Molecular aspects of fever and hyperthermia. Immunol Allergy Clin North Am 2009;29:229–45
  • Roth J, Rummel C, Barth SW, Gerstberger R, Hubschle T. Molecular aspects of fever and hyperthermia. Neurol Clin 2006;24:421–39
  • Jiang Q, DeTolla L, van Rooijen N, Singh IS, Fitzgerald B, Lipsky MM, et al. Febrile-range temperature modifies early systemic tumor necrosis factor alpha expression in mice challenged with bacterial endotoxin. Infect Immun 1999;67:1539–46
  • Rice P, Martin E, He JR, Frank M, DeTolla L, Hester L, et al. Febrile-range hyperthermia augments neutrophil accumulation and enhances lung injury in experimental Gram-negative bacterial pneumonia. J Immunol 2005;174:3676–85
  • Gupta A, Cooper ZA, Tulapurkar ME, Potla R, Maity T, Hasday JD, et al. Toll-like receptor agonists and febrile range hyperthermia synergize to induce heat shock protein 70 expression and extracellular release. J Biol Chem 2013;288:2756–66
  • Hasday JD, Fairchild KD, Shanholtz C. The role of fever in the infected host. Microbes Infect 2000;2:1891–904
  • Bernheim HA, Bodel PT, Askenase PW, Atkins E. Effects of fever on host defense mechanisms after infection in the lizard Dipsosaurus dorsalis. Br J Exp Pathol 1978;59:76–84
  • Covert JB, Reynolds WW. Survival value of fever in fish. Nature 1977;267:43–5
  • Jiang Q, Cross AS, Singh IS, Chen TT, Viscardi RM, Hasday JD. Febrile core temperature is essential for optimal host defense in bacterial peritonitis. Infect Immun 2000;68:1265–70
  • Mackowiak PA, Browne RH, Southern PM, Jr., Smith JW. Polymicrobial sepsis: An analysis of 184 cases using log linear models. Am J Med Sci 1980;280:73–80
  • Hoefs JC, Canawati HN, Sapico FL, Hopkins RR, Weiner J, Montgomerie JZ. Spontaneous bacterial peritonitis. Hepatology 1982;2:399–407
  • Ahkee S, Srinath L, Ramirez J. Community-acquired pneumonia in the elderly: Association of mortality with lack of fever and leukocytosis. South Med J 1997;90:296–8
  • Sareh H, Tulapurkar ME, Shah NG, Singh IS, Hasday JD. Response of mice to continuous 5-day passive hyperthermia resembles human heat acclimation. Cell Stress Chaperones 2011;16:297–307
  • Tulapurkar ME, Hasday JD, Singh IS. Prolonged exposure to hyperthermic stress augments neutrophil recruitment to lung during the post-exposure recovery period. Int J Hyperthermia 2011;27:717–25
  • Lipke AB, Matute-Bello G, Herrero R, Kurahashi K, Wong VA, Mongovin SM, et al. Febrile-range hyperthermia augments lipopolysaccharide-induced lung injury by a mechanism of enhanced alveolar epithelial apoptosis. J Immunol 2010;184:3801–13
  • Lipke AB, Matute-Bello G, Herrero R, Wong VA, Mongovin SM, Martin TR. Death receptors mediate the adverse effects of febrile-range hyperthermia on the outcome of lipopolysaccharide-induced lung injury. Am J Physiol Lung Cell Mol Physiol 2011;301:L60–70
  • Hasday JD, Garrison A, Singh IS, Standiford T, Ellis GS, Rao S, et al. Febrile-range hyperthermia augments pulmonary neutrophil recruitment and amplifies pulmonary oxygen toxicity. Am J Pathol 2003;162:2005–17
  • D'Alessio FR, Tsushima K, Aggarwal NR, West EE, Willett MH, Britos MF, et al. CD4+CD25+Foxp3+ Tregs resolve experimental lung injury in mice and are present in humans with acute lung injury. J Clin Invest 2009;119:2898–913
  • Brenu EW, Staines DR, Tajouri L, Huth T, Ashton KJ, Marshall-Gradisnik SM. Heat shock proteins and regulatory T cells. Autoimmune Dis 2013;2013:813256
  • Mace TA, Zhong L, Kokolus KM, Repasky EA. Effector CD8+ T cell IFN-gamma production and cytotoxicity are enhanced by mild hyperthermia. Int J Hyperthermia 2012;28:9–18
  • Wachstein J, Tischer S, Figueiredo C, Limbourg A, Falk C, Immenschuh S, et al. HSP70 enhances immunosuppressive function of CD4(+)CD25(+)FoxP3(+) T regulatory cells and cytotoxicity in CD4(+)CD25(-) T cells. PLoS One 2012;7:e51747
  • Chen Q, Fisher DT, Clancy KA, Gauguet JM, Wang WC, Unger E, et al. Fever-range thermal stress promotes lymphocyte trafficking across high endothelial venules via an interleukin 6 trans-signaling mechanism. Nat Immunol 2006;7:1299–308
  • Ellis GS, Carlson DE, Hester L, He JR, Bagby GJ, Singh IS, et al. G-CSF, but not corticosterone, mediates circulating neutrophilia induced by febrile-range hyperthermia. J Appl Physiol 2005;98:1799–804
  • Tulapurkar ME, Almutairy EA, Shah NG, He JR, Puche AC, Shapiro P, et al. Febrile-range hyperthermia modifies endothelial and neutrophilic functions to promote extravasation. Am J Respir Cell Mol Biol 2012;46:807–14
  • Hasday JD, Bannerman D, Sakarya S, Cross AS, Singh IS, Howard D, et al. Exposure to febrile temperature modifies endothelial cell response to tumor necrosis factor-alpha. J Appl Physiol 2001;90:90–8
  • Nagarsekar A, Greenberg RS, Shah NG, Singh IS, Hasday JD. Febrile-range hyperthermia accelerates caspase-dependent apoptosis in human neutrophils. J Immunol 2008;181:2636–43
  • Nagarsekar A, Tulapurkar ME, Singh IS, Atamas SP, Shah NG, Hasday JD. Hyperthermia promotes and prevents respiratory epithelial apoptosis through distinct mechanisms. Am J Respir Cell Mol Biol 2012;47:824–33
  • Lee CT, Repasky EA. Opposing roles for heat and heat shock proteins in macrophage functions during inflammation: A function of cell activation state? Front Immunol 2012;3:140
  • Ostberg JR, Taylor SL, Baumann H, Repasky EA. Regulatory effects of fever-range whole-body hyperthermia on the LPS-induced acute inflammatory response. J Leukoc Biol 2000;68:815–20
  • Evans SS, Wang WC, Bain MD, Burd R, Ostberg JR, Repasky EA. Fever-range hyperthermia dynamically regulates lymphocyte delivery to high endothelial venules. Blood 2001;97:2727–33
  • Wang WC, Goldman LM, Schleider DM, Appenheimer MM, Subjeck JR, Repasky EA, Evans SS. Fever-range hyperthermia enhances L-selectin-dependent adhesion of lymphocytes to vascular endothelium. J Immunol 1998;160:961–9
  • Fisher DT, Chen Q, Skitzki JJ, Muhitch JB, Zhou L, Appenheimer MM, et al. IL-6 trans-signaling licenses mouse and human tumor microvascular gateways for trafficking of cytotoxic T cells. J Clin Invest 2011;121:3846–59
  • Singh V, Aballay A. Heat shock and genetic activation of HSF-1 enhance immunity to bacteria. Cell Cycle 2006;5:2443–6
  • Xiao X, Zuo X, Davis AA, McMillan DR, Curry BB, Richardson JA, et al. HSF1 is required for extra-embryonic development, postnatal growth and protection during inflammatory responses in mice. Embo J 1999;18:5943–52
  • Westwood JT, Clos J, Wu C. Stress-induced oligomerization and chromosomal relocalization of heat-shock factor. Nature 1991;353:822–7
  • Trinklein ND, Chen WC, Kingston RE, Myers RM. Transcriptional regulation and binding of heat shock factor 1 and heat shock factor 2 to 32 human heat shock genes during thermal stress and differentiation. Cell Stress Chaperones 2004;9:21–8
  • Nagarsekar A, Hasday JD, Singh IS. CXC chemokines: A new family of heat-shock proteins? Immunol Invest 2005;34:381–98
  • Maity TK, Henry MM, Tulapurkar ME, Shah NG, Hasday JD, Singh IS. Distinct, gene-specific effect of heat shock on heat shock factor-1 recruitment and gene expression of CXC chemokine genes. Cytokine 2011;54:61–7
  • Singh IS, Gupta A, Nagarsekar A, Cooper Z, Manka C, Hester L, et al. Heat shock co-activates interleukin-8 transcription. Am J Respir Cell Mol Biol 2008;39:235–42
  • Dinh HK, Zhao B, Schuschereba ST, Merrill G, Bowman PD. Gene expression profiling of the response to thermal injury in human cells. Physiol Genomics 2001;7:3–13
  • Murray JI, Whitfield ML, Trinklein ND, Myers RM, Brown PO, Botstein D. Diverse and specific gene expression responses to stresses in cultured human cells. Mol Biol Cell 2004;15:2361–74
  • Sonna LA, Gaffin SL, Pratt RE, Cullivan ML, Angel KC, Lilly CM. Effect of acute heat shock on gene expression by human peripheral blood mononuclear cells. J Appl Physiol 2002;92:2208–20
  • Mendillo ML, Santagata S, Koeva M, Bell GW, Hu R, Tamimi RM, et al. HSF1 drives a transcriptional program distinct from heat shock to support highly malignant human cancers. Cell 2012;150:549–62
  • Christians E, Davis AA, Thomas SD, Benjamin IJ. Maternal effect of Hsf1 on reproductive success. Nature 2000;407:693–4
  • Izu H, Inouye S, Fujimoto M, Shiraishi K, Naito K, Nakai A. Heat shock transcription factor 1 is involved in quality-control mechanisms in male germ cells. Biol Reprod 2004;70:18–24
  • Pirkkala L, Alastalo TP, Zuo X, Benjamin IJ, Sistonen L. Disruption of heat shock factor 1 reveals an essential role in the ubiquitin proteolytic pathway. Mol Cell Biol 2000;20:2670–5
  • Santos SD, Saraiva MJ. Enlarged ventricles, astrogliosis and neurodegeneration in heat shock factor 1 null mouse brain. Neuroscience 2004;126:657–63
  • Takaki E, Fujimoto M, Nakahari T, Yonemura S, Miyata Y, Hayashida N, et al. Heat shock transcription factor 1 is required for maintenance of ciliary beating in mice. J Biol Chem 2007;282:37285–92
  • Takaki E, Fujimoto M, Sugahara K, Nakahari T, Yonemura S, Tanaka Y, et al. Maintenance of olfactory neurogenesis requires HSF1, a major heat shock transcription factor in mice. J Biol Chem 2006;281:4931–7
  • Dai C, Whitesell L, Rogers AB, Lindquist S. Heat shock factor 1 is a powerful multifaceted modifier of carcinogenesis. Cell 2007;130:1005–18
  • Xie Y, Chen C, Stevenson MA, Auron PE, Calderwood SK. Heat shock factor 1 represses transcription of the IL-1beta gene through physical interaction with the nuclear factor of interleukin 6. J Biol Chem 2002;277:11802–10
  • Xie Y, Zhong R, Chen C, Calderwood SK. Heat shock factor 1 contains two functional domains that mediate transcriptional repression of the c-fos and c-fms genes. J Biol Chem 2003;278:4687–98
  • Chen C, Xie Y, Stevenson MA, Auron PE, Calderwood SK. Heat shock factor 1 represses Ras-induced transcriptional activation of the c-fos gene. J Biol Chem 1997;272:26803–6
  • Cahill CM, Waterman WR, Xie Y, Auron PE, Calderwood SK. Transcriptional repression of the prointerleukin 1beta gene by heat shock factor 1. J Biol Chem 1996;271:24874–9
  • Singh IS, He JR, Hester L, Fenton MJ, Hasday JD. Bacterial endotoxin modifies heat shock factor-1 activity in RAW 264.7 cells: Implications for TNF-alpha regulation during exposure to febrile range temperatures. J Endotoxin Res 2004;10:175–84
  • Singh IS, He JR, Calderwood S, Hasday JD. A high affinity HSF-1 binding site in the 5'-untranslated region of the murine tumor necrosis factor-alpha gene is a transcriptional repressor. J Biol Chem 2002;277:4981–8
  • Cooper ZA, Singh IS, Hasday JD. Febrile range temperature represses TNF-alpha gene expression in LPS-stimulated macrophages by selectively blocking recruitment of Sp1 to the TNF-alpha promoter. Cell Stress Chaperones 2010;15:665–73
  • Cooper ZA, Ghosh A, Gupta A, Maity T, Benjamin IJ, Vogel SN, et al. Febrile-range temperature modifies cytokine gene expression in LPS-stimulated macrophages by differentially modifying NF-{kappa}B recruitment to cytokine gene promoters. Am J Physiol Cell Physiol 2010;298:C171–81
  • Wang J, He H, Yu L, Xia HH, Lin MC, Gu Q, et al. HSF1 down-regulates XAF1 through transcriptional regulation. J Biol Chem 2006;281:2451–9
  • Goldring CE, Reveneau S, Chantome A, Pance A, Fleury C, Hume DA, et al. Heat shock enhances transcriptional activation of the murine-inducible nitric oxide synthase gene. FASEB J 2000;14:2393–5
  • Inouye S, Fujimoto M, Nakamura T, Takaki E, Hayashida N, Hai T, et al. Heat shock transcription factor 1 opens chromatin structure of interleukin-6 promoter to facilitate binding of an activator or a repressor. J Biol Chem 2007;282:33210–17
  • Rossi A, Trotta E, Brandi R, Arisi I, Coccia M, Santoro MG. AIRAP, a new human heat shock gene regulated by heat shock factor 1. J Biol Chem 2010;285:13607–15
  • Rossi A, Coccia M, Trotta E, Angelini M, Santoro MG. Regulation of cyclooxygenase-2 expression by heat: A novel aspect of heat shock factor 1 function in human cells. PLoS One 2012;7:e31304
  • Skaggs HS, Xing H, Wilkerson DC, Murphy LA, Hong Y, Mayhew CN, et al. HSF1-TPR interaction facilitates export of stress-induced HSP70 mRNA. J Biol Chem 2007;282:33902–7
  • Huang J, Nueda A, Yoo S, Dynan WS. Heat shock transcription factor 1 binds selectively in vitro to Ku protein and the catalytic subunit of the DNA-dependent protein kinase. J Biol Chem 1997;272:26009–16
  • Xie Y, Chen C, Stevenson MA, Hume DA, Auron PE, Calderwood SK. NF-IL6 and HSF1 have mutually antagonistic effects on transcription in monocytic cells. Biochem Biophys Res Commun 2002;291:1071–80
  • Singh U, Bongcam-Rudloff E, Westermark B. A DNA sequence directed mutual transcription regulation of HSF1 and NFIX involves novel heat sensitive protein interactions. PLoS One 2009;4:e5050
  • Yuan CX, Gurley WB. Potential targets for HSF1 within the preinitiation complex. Cell Stress Chaperones 2000;5:229–42
  • Lee YJ, Lee HJ, Lee JS, Jeoung D, Kang CM, Bae S, et al. A novel function for HSF1-induced mitotic exit failure and genomic instability through direct interaction between HSF1 and Cdc20. Oncogene 2008;27:2999–3009
  • Charette SJ, Lavoie JN, Lambert H, Landry J. Inhibition of Daxx-mediated apoptosis by heat shock protein 27. Mol Cell Biol 2000;20:7602–12
  • Singhal SS, Yadav S, Drake K, Singhal J, Awasthi S. Hsf-1 and POB1 induce drug sensitivity and apoptosis by inhibiting Ralbp1. J Biol Chem 2008;283:19714–29
  • Li QL, Ju ZH, Huang JM, Li JB, Li RL, Hou MH, et al. Two novel SNPs in HSF1 gene are associated with thermal tolerance traits in Chinese Holstein cattle. DNA Cell Biol 2011;30:247–54
  • Bridges TM, Singh IS, Liggett SB, Hasday JD. Polymorphisms of heat shock factor-1: Genetic divesity in an evolutionarily conserved, multifunctional regulator gene. Am J Respir Crit Care Med 2011;183:A6018
  • Barenboim M, Zoltick BJ, Guo Y, Weinberger DR. MicroSNiPer: A web tool for prediction of SNP effects on putative microRNA targets. Hum Mutat 2010;31:1223–32
  • Gruber AR, Lorenz R, Bernhart SH, Neubock R, Hofacker IL. The Vienna RNA websuite. Nucleic Acids Res 2008;36:W70–4
  • Hashiguchi N, Ogura H, Tanaka H, Koh T, Nakamori Y, Noborio M, et al. Enhanced expression of heat shock proteins in activated polymorphonuclear leukocytes in patients with sepsis. J Trauma 2001;51:1104–9
  • Delogu G, Lo Bosco L, Marandola M, Famularo G, Lenti L, Ippoliti F, et al. Heat shock protein (HSP70) expression in septic patients. J Crit Care 1997;12:188–92
  • Wheeler DS, Fisher LE, Jr., Catravas JD, Jacobs BR, Carcillo JA, Wong HR. Extracellular hsp70 levels in children with septic shock. Pediatr Crit Care Med 2005;6:308–11
  • Njemini R, Lambert M, Demanet C, Mets T. Elevated serum heat-shock protein 70 levels in patients with acute infection: Use of an optimized enzyme-linked immunosorbent assay. Scand J Immunol 2003;58:664–9
  • Njemini R, Bautmans I, Onyema OO, Van Puyvelde K, Demanet C, Mets T. Circulating heat shock protein 70 in health, aging and disease. BMC Immunol 2011;12:24
  • Stulik J, Hernychova L, Macela A, Krocova Z, Kroca M. Production of stress-inducible form of heat-shock protein 70 in mouse peritoneal adherent cells after in vivo infection by Francisella tularensis. Folia Microbiol (Praha) 1999;44:306–10
  • Martinez J, Perez-Serrano J, Bernadina WE, Rodriguez-Caabeiro F. Shock response induced in rat brain and spleen during primary infection with Trichinella spiralis larvae. Parasitology 1999;118:605–13
  • Singleton KD, Serkova N, Beckey VE, Wischmeyer PE. Glutamine attenuates lung injury and improves survival after sepsis: Role of enhanced heat shock protein expression. Crit Care Med 2005;33:1206–13
  • Weiss YG, Bouwman A, Gehan B, Schears G, Raj N, Deutschman CS. Cecal ligation and double puncture impairs heat shock protein 70 (HSP-70) expression in the lungs of rats. Shock 2000;13:19–23
  • Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F, Edberg S, Medzhitov R. Recognition of commensal microflora by Toll-like receptors is required for intestinal homeostasis. Cell 2004;118:229–41
  • Kotsiopriftis M, Tanner JE, Alfieri C. Heat shock protein 90 expression in Epstein-Barr virus-infected B cells promotes gammadelta T-cell proliferation in vitro. J Virol 2005;79:7255–61
  • Brasier AR, Spratt H, Wu Z, Boldogh I, Zhang Y, Garofalo RP, et al. Nuclear heat shock response and novel nuclear domain 10 reorganization in respiratory syncytial virus-infected a549 cells identified by high-resolution two-dimensional gel electrophoresis. J Virol 2004;78:11461–76
  • Melcher A, Murphy S, Vile R. Heat shock protein expression in target cells infected with low levels of replication-competent virus contributes to the immunogenicity of adenoviral vectors. Hum Gene Ther 1999;10:1431–42
  • O'Reilly DA, Roberts JR, Cartmell MT, Demaine AG, Kingsnorth AN. Heat shock factor-1 and nuclear factor-kappaB are systemically activated in human acute pancreatitis. JOP 2006;7:174–84
  • Ethridge RT, Ehlers RA, Hellmich MR, Rajaraman S, Evers BM. Acute pancreatitis results in induction of heat shock proteins 70 and 27 and heat shock factor-1. Pancreas 2000;21:248–56
  • McGrath LB, Locke M, Cane M, Chen C, Ianuzzo CD. Heat shock protein (HSP 72) expression in patients undergoing cardiac operations. J Thorac Cardiovasc Surg 1995;109:370–6
  • Taggart DP, Bakkenist CJ, Biddolph SC, Graham AK, McGee JO. Induction of myocardial heat shock protein 70 during cardiac surgery. J Pathol 1997;182:362–6
  • Calderwood SK, Mambula SS, Gray PJ, Jr., Theriault JR. Extracellular heat shock proteins in cell signaling. FEBS Lett 2007;581:3689–94
  • De Nardo D, Masendycz P, Ho S, Cross M, Fleetwood AJ, Reynolds EC, et al. A central role for the Hsp90.Cdc37 molecular chaperone module in interleukin-1 receptor-associated-kinase-dependent signaling by toll-like receptors. J Biol Chem 2005;280:9813–22
  • Vabulas RM, Ahmad-Nejad P, Ghose S, Kirschning CJ, Issels RD, Wagner H. HSP70 as endogenous stimulus of the Toll/interleukin-1 receptor signal pathway. J Biol Chem 2002;277:15107–12
  • Tsan MF, Gao B. Heat shock proteins and immune system. J Leukoc Biol 2009;85:905–10
  • Gao B, Tsan MF. Endotoxin contamination in recombinant human heat shock protein 70 (Hsp70) preparation is responsible for the induction of tumor necrosis factor alpha release by murine macrophages. J Biol Chem 2003;278:174–9
  • Zheng H, Nagaraja GM, Kaur P, Asea EE, Asea A. Chaperokine function of recombinant Hsp72 produced in insect cells using a baculovirus expression system is retained. J Biol Chem 2010;285:349–56
  • Wang R, Kovalchin JT, Muhlenkamp P, Chandawarkar RY. Exogenous heat shock protein 70 binds macrophage lipid raft microdomain and stimulates phagocytosis, processing, and MHC-II presentation of antigens. Blood 2006;107:1636–42
  • Vega VL, Rodriguez-Silva M, Frey T, Gehrmann M, Diaz JC, Steinem C, et al. Hsp70 translocates into the plasma membrane after stress and is released into the extracellular environment in a membrane-associated form that activates macrophages. J Immunol 2008;180:4299–307
  • Asea A. Heat shock proteins and toll-like receptors. Handb Exp Pharmacol 2008;183:111–27
  • Aneja R, Odoms K, Dunsmore K, Shanley TP, Wong HR. Extracellular heat shock protein-70 induces endotoxin tolerance in THP-1 cells. J Immunol 2006;177:7184–92
  • Stocki P, Dickinson AM. The immunosuppressive activity of heat shock protein 70. Autoimmune Dis 2012;2012:617213
  • Pockley AG, Muthana M, Calderwood SK. The dual immunoregulatory roles of stress proteins. Trends Biochem Sci 2008;33:71–9
  • Torigoe T, Tamura Y, Sato N. Heat shock proteins and immunity: Application of hyperthermia for immunomodulation. Int J Hyperthermia 2009;25:610–16
  • Borges TJ, Wieten L, van Herwijnen MJ, Broere F, van der Zee R, Bonorino C, et al. The anti-inflammatory mechanisms of Hsp70. Front Immunol 2012;3:95
  • Hunter-Lavin C, Davies EL, Bacelar MM, Marshall MJ, Andrew SM, Williams JH. Hsp70 release from peripheral blood mononuclear cells. Biochem Biophys Res Commun 2004;324:511–17
  • Hightower LE, Guidon PT, Jr. Selective release from cultured mammalian cells of heat-shock (stress) proteins that resemble glia-axon transfer proteins. J Cell Physiol 1989;138:257–66
  • Basu S, Binder RJ, Suto R, Anderson KM, Srivastava PK. Necrotic but not apoptotic cell death releases heat shock proteins, which deliver a partial maturation signal to dendritic cells and activate the NF-kappa B pathway. Int Immunol 2000;12:1539–46
  • Mambula SS, Calderwood SK. Heat induced release of Hsp70 from prostate carcinoma cells involves both active secretion and passive release from necrotic cells. Int J Hyperthermia 2006;22:575–85
  • Evdonin AL, Martynova MG, Bystrova OA, Guzhova IV, Margulis BA, Medvedeva ND. The release of Hsp70 from A431 carcinoma cells is mediated by secretory-like granules. Eur J Cell Biol 2006;85:443–55
  • Mambula SS, Stevenson MA, Ogawa K, Calderwood SK. Mechanisms for Hsp70 secretion: crossing membranes without a leader. Methods 2007;43:168–75
  • Mambula SS, Calderwood SK. Heat shock protein 70 is secreted from tumor cells by a nonclassical pathway involving lysosomal endosomes. J Immunol 2006;177:7849–57
  • Edelman DA, Jiang Y, Tyburski JG, Wilson RF, Steffes CP. Lipopolysaccharide up-regulates heat shock protein expression in rat lung pericytes. J Surg Res 2007;140:171–6
  • Hirsh MI, Hashiguchi N, Chen Y, Yip L, Junger WG. Surface expression of HSP72 by LPS-stimulated neutrophils facilitates gammadeltaT cell-mediated killing. Eur J Immunol 2006;36:712–21
  • Unoshima M, Nishizono A, Takita-Sonoda Y, Iwasaka H, Noguchi T. Effects of zinc acetate on splenocytes of endotoxemic mice: Enhanced immune response, reduced apoptosis, and increased expression of heat shock protein 70. J Lab Clin Med 2001;137:28–37
  • Ofenstein JP, Heidemann S, Juett-Wilstermann A, Sarnaik A. Expression of stress proteins HSP 72 & HSP 32 in response to endotoxemia. Ann Clin Lab Sci 2000;30:92–8
  • Flohe S, Dominguez Fernandez E, Ackermann M, Hirsch T, Borgermann J, Schade FU. Endotoxin tolerance in rats: Expression of TNF-alpha, IL-6, IL-10, VCAM-1 and HSP 70 in lung and liver during endotoxin shock. Cytokine 1999;11:796–804
  • Murapa P, Gandhapudi S, Skaggs HS, Sarge KD, Woodward JG. Physiological fever temperature induces a protective stress response in T lymphocytes mediated by heat shock factor-1 (HSF1). J Immunol 2007;179:8305–12
  • Sarge KD, Bray AE, Goodson ML. Altered stress response in testis. Nature 1995;374:126
  • Sarge KD. Male germ cell-specific alteration in temperature set point of the cellular stress response. J Biol Chem 1995;270:18745–8

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.